Skip to main content

Combining DNA methylation and RNA sequencing data of cancer for supervised knowledge extraction

Abstract

Background

In the Next Generation Sequencing (NGS) era a large amount of biological data is being sequenced, analyzed, and stored in many public databases, whose interoperability is often required to allow an enhanced accessibility. The combination of heterogeneous NGS genomic data is an open challenge: the analysis of data from different experiments is a fundamental practice for the study of diseases. In this work, we propose to combine DNA methylation and RNA sequencing NGS experiments at gene level for supervised knowledge extraction in cancer.

Methods

We retrieve DNA methylation and RNA sequencing datasets from The Cancer Genome Atlas (TCGA), focusing on the Breast Invasive Carcinoma (BRCA), the Thyroid Carcinoma (THCA), and the Kidney Renal Papillary Cell Carcinoma (KIRP). We combine the RNA sequencing gene expression values with the gene methylation quantity, as a new measure that we define for representing the methylation quantity associated to a gene. Additionally, we propose to analyze the combined data through tree- and rule-based classification algorithms (C4.5, Random Forest, RIPPER, and CAMUR).

Results

We extract more than 15,000 classification models (composed of gene sets), which allow to distinguish the tumoral samples from the normal ones with an average accuracy of 95%. From the integrated experiments we obtain about 5000 classification models that consider both the gene measures related to the RNA sequencing and the DNA methylation experiments.

Conclusions

We compare the sets of genes obtained from the classifications on RNA sequencing and DNA methylation data with the genes obtained from the integration of the two experiments. The comparison results in several genes that are in common among the single experiments and the integrated ones (733 for BRCA, 35 for KIRP, and 861 for THCA) and 509 genes that are in common among the different experiments. Finally, we investigate the possible relationships among the different analyzed tumors by extracting a core set of 13 genes that appear in all tumors. A preliminary functional analysis confirms the relation of part of those genes (5 out of 13 and 279 out of 509) with cancer, suggesting to focus further studies on the new individuated ones.

Peer Review reports

Introduction

Next Generation Sequencing (NGS) techniques have revolutionized the sequencing of genomes, producing large quantities of DNA and RNA data [14]. This abundance of data allows us to perform analyses on the genetic makeup of human subjects, studying the predisposition to diseases like cancer [58]. NGS techniques are not only applied to DNA sequencing [9], but also to other types of experiments, e.g.: transcriptome profiling (RNA sequencing) [10, 11], microRNA sequencing (miRNA-seq) [12], protein-DNA interactions (Chip-Seq) [13], identification of Copy Number Variation (CNV) [14], and characterization of the epigenome or chemical changes in the DNA (DNA methylation) [1517].

In this work, we are going to focus on DNA methylation and RNA sequencing, as these two NGS experiments have been proven to play an important role in knowledge discovery in cancer [1825].

DNA methylation is one of the most studied epigenetic changes in human cells. The changes in DNA methylation patterns are crucial in the development of diseases and in many forms of cancer [2631]. Most NGS methods are based on bisulfite conversion to determine the percentage of methylated cytosines in a CpG island. This measure is called beta value [32], and is defined as the ratio between the methylated allele intensity and the overall intensity. For more details about the DNA methylation experimental techniques the reader may refer to [17, 33].

RNA sequencing is a next generation sequencing technique for the analysis of the transcriptome and its quantification. Four main methods for measuring gene expression are used in practice: i) Reads Per Kilobase per Million mapped reads (RPKM)[10]; ii) Fragments Per Kilobase per Million mapped (FPKM) [34]; iii) RNA-Seq by Expectation-Maximization (RSEM) [11, 35]; iv) Transcripts Per Kilobase Million (TPM) [36]. For further details about RNA sequencing, we point the reader to [37], where the authors perform a comprehensive overview of this NGS technique.

In this work, we define NGS data the information extracted from a NGS experiment (i.e., Chip-sequencing, DNA methylation, DNA sequencing, RNA sequencing), e.g., the counts of the reads that map on given list of genes in RNA sequencing. We define NGS meta data the information related to the NGS experiment and the sequenced tissue, e.g., the tissues status (tumoral, normal), or the sequencing depth. We define NGS data integration the procedure of joining different experiments (possibly extracted from heterogeneous databases) sharing common features (e.g., same disease / patient under study) in order to extract knowledge. The aim of integration is to aggregate genomic data in an unique schema that provides querying capabilities for retrieving data from a multitude of heterogeneous experiments and databases. Heterogeneous data are the first problem of NGS, because the structure of data is different in diverse experiments and can be different in diverse databases. Therefore, the term integration in NGS data can have different meanings [38]. On one hand, we consider integration for a need to have a uniform language that facilitates the access to different genomic databases. On the other hand data heterogeneity is caused by the experiment types and by the information that they bring. It is worth noting that dis-uniformity of the data schema is present not only when considering different databases, but also when dealing with a single one. We distinguish four conditions, where NGS data integration can be performed: (i) different databases represent the same NGS experiment (e.g. RNA-Seq) with different data schemas; (ii) different experiments (e.g., DNA methylation and RNA-Seq) in distinct databases; in this case there are two different data schemas, because the experiments need a different representation, but no standardization of the schemas is defined that allows the access to these experiments; (iii) the same problem exists even in the same databases, which contains different experiments and different data representation schemas. Finally, we consider an ideal case (iv) where a previously defined schema standardization allows to integrate different experiments that come from different databases or from the same database, and it allows also to provide interoperability between the same experiments but with different schemas. An example of this type of standardization is provided by [39] with the Genomic Data Model (GDM) that supports many NGS formats.

In order to have access to the right resources, it is necessary to define standard schemas of these data to avoid redundant information overlaps. Several efforts have been made on NGS data formats and standards. The authors of [40] provide the reader with an overview of the most widespread data formats for NGS and describe a set of standardization approaches for them. In [41] the NCBI Entrez search and retrieval system used at the National Center for Biotechnology Information to access distributed heterogeneous data is described. Also the authors of [42] present a text search engine to access data resources in the European Bioinformatics Institute (EMBL-EBI) and to help understand the relationship between different data types. Other implementations for bioinformatics data integration include retrieval systems like SRS [43] and integration tools for information fusion such as BioData Server [44]. The integration of genomic data involves multiple fields, i.e., bioinformatics, statistics, data mining, and classification. But the question is, does the integration of different types of NGS experiments offer additional knowledge about a disease like cancer [45]?

In this work, we address the issue of combining RNA sequencing and DNA methylation experiments, which have different data schemas containing heterogeneous information. Our aim is to obtain a gene oriented organization of both experiments, and therefore we define a new measure on DNA methylation data called gene methylation quantity. We combine RNA sequencing and DNA methylation data of The Cancer Genome Atlas (TCGA) [46] and test our method on genomic data related to three types of cancer: Breast Cancer, Kidney Renal Carcinoma, and Thyroid Carcinoma.

Additionally, we analyze the combined data by means of supervised classification algorithms, extracting classification models, which are able to distinguish the samples in two classes (tumoral and normal) and which are composed of features that represent the genes related to the disease and the different NGS experiment.

In cancer research many computational methods deal with classification problems, e.g., disease characterization, prognosis, treatment response of patients, mutation pathogenicity, biomarker prediction, and sample malignancy. A recent effort has achieved good performance in the assignment of disease subtypes and malignancy labels to melanoma images with convolutional neural networks [47]. Further studies used typical machine learning methods [48], including Adaboost [49] and decision trees [50].

Among them, we focus on a new supervised learning method that is able to extract more knowledge in terms of classification models than state of the art ones, called Classifier with Alternative and MUltiple Rule-based models (CAMUR) [51]. CAMUR is designed to find alternative and equivalent solutions for a classification problem building multiple rule-based classification models. Standard classifiers tend to extract few rules with a small set of features for discriminating the samples, and interesting features may remain hidden from the researcher. Thanks to an iterative classification procedure based on a feature elimination technique, CAMUR finds a large number of rules related to the classes present in the dataset under study. CAMUR is based on: (i) a rule-based classifier, i.e., RIPPER (Repeated Incremental Pruning to Produce Error Reduction) [52]; (ii) an iterative feature elimination technique; (iii) a repeated classification procedure; (iv) a storage structure for the classification rules. The method calculates iteratively a rule-based classification model through the RIPPER algorithms [52], deletes iteratively the features that are present in the rules from the dataset, and performs the classification procedure again, until a stopping criterion is met, i.e., the classification performance is below a given threshold or the maximum number of iterations has been reached. CAMUR has been implemented specifically for case-control studies that aim to identify subjects by their outcome status (e.g., tumoral or normal). In these data, the features correspond to the gene expressions of the samples, the classes to the investigated diseases or conditions (e.g., tumoral, normal). The extracted knowledge by CAMUR consists in a set of rules composed of a given number of genes that might be relevant for a disease. CAMUR also includes an offline tool to analyze and to interpret the computed results. Thus the software consists of two parts: (i) The Multiple Solutions Extractor (MSE), which corresponds to the implementation of the iterative classification algorithm (i.e., for each iteration it deletes the selected features, performs the classification, and saves the extracted models); (ii) The Multiple Solutions Analyzer (MSA), a graphical tool for analyzing and interpreting the obtained results. CAMUR is available at http://dmb.iasi.cnr.it/camur.php as stand alone software; for a comprehensive description we point the reader to [51].

In this work, thanks to the application of machine learning algorithms, we show the advantage of combining DNA methylation and RNA sequencing data, i.e., the increase of extracted knowledge resulting in combinations of genes from both experimental strategies. Finally, we study the three types of cancer and identify sets of relevant genes. The intersection of them results in a smaller set of genes that should be considered for further investigation.

Methods

In this section, we discuss the methods used to combine the genomic experiments (RNA sequencing, DNA methylation) and the classification algorithms used to extract knowledge from them. We start by describing the source where we extract the data.

Data source: the Cancer Genome Atlas

The Cancer Genome Atlas (TCGA) [46] is a project that aims to create a major repository for cancer, including NGS experiments, to improve the ability to diagnose, treat and prevent cancer through a better understanding of the genetic basis of this disease. The TCGA database contains the genomic characterization and analysis of 33 types of cancer. Tissue samples are processed through different types of techniques such as gene expression profiling (i.e., RNA sequencing and microarrays); profiling of methylated DNA (i.e., DNA methylation obtained both with NGS techniques and microarrays); profiling of microRNA (i.e., miRNA sequencing); whole genome sequecing (i.e., DNA sequencing). We rely on the latest TCGA data release available at The Genomic Data Commons platform (http://gdc.cancer.gov/).

In TCGA each tissue of DNA methylation is represented with a list of following fields: gene symbol, chromosome and genomic coordinates (where the methylation occurs), and its beta value (methylation values). RNA sequencing data instead contains information on the RSEM values [11] measured on the considered genes. It is worth noting that our approach handles gene expression data of RNA sequencing, which has been previously normalized, DNA methylation data containing the beta value, and can be used to treat also DNA methylation and RNA sequencing data of different pathologies.

Data processing and combination

We create data matrices of RNA sequencing and DNA methylation experiments in the following way. Consider n samples (tissues) each one with m features (genes) and a class label (condition), which indicates whether the sample is normal or tumoral. A data matrix is composed by n vectors as Fi = (fi,1,fi,2,...,fi,m,fi,c), which represent sample i, where fi,j \(\mathbb {R}\); i=1,...,n; j=1,...,m; fi,c {normal,tumoral}. When considering RNA sequencing, the rows represent the samples, the columns the genes (except the last that represents the class labels) and the items of the matrix contain the RSEM gene expression values for each gene. The structure of this matrix is shown in Table 1. When considering DNA methylation, the corresponding matrix is composed by the rows that represent the samples, the columns that represent the genes, while the items contain a new measure that represent the quantity of methylation associates to each gene and that is explained in the following. Indeed, for DNA methylation TCGA encloses the beta values for each methylated site, so each sample has s methylated sites, l of them belonging to a given gene. For aggregating the methylation quantity at gene level, we consider the sum of the beta values as a measure of the overall intensity of the methylation on a gene. Let aijh be the methylation quantity associated to the sample i with i=1,..,n, to the gene j with j=1,....m, and to the methylated site h with h=1,..,l. Then we have \(b_{i,j} = \sum _{h=1}^{l} a_{ijh}, \forall i,j\). In the following, we refer to this new measure as gene methylation quantity. It is worth noting that we consider the beta values of CpG sites with a related gene symbol, i.e., the symbol of the gene where the methylation occurs. If a methylation occurs on other genomic regions it is not considered in our data processing procedure, whose aim is to provide a gene oriented data organization. In Table 2 we show the structure of the DNA methylation matrix. A software tool, which performs the data extraction and the creation of the matrices, is freely available at http://bioinf.iasi.cnr.it/genint. The flowchart that reports the computational steps of the software is depicted in Fig. 1.

Fig. 1
figure 1

The flowchart of the computational steps for creating the RNA sequencing and DNA methylation matrices. The first step represents the parsing of the input dataset of TCGA. The samples are read for the extraction of the features (genes) and their related values, which are the gene expression measures in case of RNA sequencing, or the methylation quantities for each gene in case of DNA methylation. Subsequently, the samples and the related gene-value pairs are unified in a single file. From this file the header (columns) and the values (rows) of the matrix are created. In the final matrix (comma separated values format), the header reports all the genes, while the rows are identified by the sample id and report the related values

Table 1 Structure of the RNA sequencing matrix
Table 2 Structure of the DNA methylation matrix

In order to perform our analysis on both gene oriented measures (RSEM for RNA sequencing and gene methylation quantity for DNA methylation) at the same time, we propose a combination of these two experiments by applying an intersection of the matrices on common sample IDs and a union of those not in common (this result trace over the full outer join in SQL language), keeping both experimental data and performing a union of the genes that are present in RNA sequencing and DNA methylation as features. The resulting matrix is shown in Table 3. Let i be the i-th sample, j the j-th gene, with i=1,..,n and j=1,..,m in case of a gene of the DNA methylation experiment, j=m+1,....,z in case of a gene of the RNA sequencing experiment. Furthermore, we have bi,j and vali,j \(\{\mathbb {R},?\}\), j,i, where the “?” symbol means that there is no value associated to the gene j for the sample i. We release also a software that is able to perform the combination of the experiments, available at http://bioinf.iasi.cnr.it/genint. Finally, we report the main steps of the procedure in Fig. 2.

Fig. 2
figure 2

Flowchart for creating the combined matrices. Firstly, a parser reads the DNA methylation and RNA sequencing matrices in input (computed as described in Fig. 1), and sends the next elaborations to two distinct processes. A step is responsible of the creation of the full header of the combined matrix with all the genes, of both the DNA methylation and RNA sequencing. The other step takes the parsed sample IDs to modify the identification of the sample (TCGA barcode) deleting the details of the performed experiments. After the creation of the sample IDs, the join step follows: the initials matrices are joined on the modified IDs and the new rows of the matrix are created, including both gene expression and gene methylation quantity. The join defines the rows with the values of the two experiments (on which the join is made because the sample id is present in both input matrices), and also the rows with values of only one experiment (if the sample is not available in both input matrices of DNA methylation and RNA sequencing)

Table 3 Structure of the combined matrix

Analysis method

The classification that we perform has the objective of being able to determine a set of rules for each type of tumor (composed of the genes and their related values), which can define if a tissue is in tumoral or in normal condition.

It is worth noting that also in previous works [5358] DNA methylation has been used to classify data samples (and patients) of cancer, but only a subset of single methylated sites have been used as features. Recently, the authors of [59] perform the classification task by considering all the single methylated sites in the genome with big data techniques. Conversely, we use the previously defined gene methylation quantity and not the single methylated sites. Also gene expression data of RNA sequencing has been widely used for cancer classification [51, 6066] and proven to be effective in distinguishing normal from tumoral samples. Our aim is to combine both information in order to extract a wider knowledge and to better focus on those genes that are related to the disease.

For performing the task of knowledge discovery, we used four different classification algorithms that are briefly described in the following.

C4.5

C4.5 [67] is an algorithm for the generation of decision trees used for classification. The algorithm takes as input a set of classified data (training set) and the output is composed by leaf nodes of the tree, which define the belonging to a class attribute. Indeed, the path from the root to a specific leaf means that all the predicates applied to the features of the sample are verified. The validity of the tree is verified on a set of labeled samples (test set), but whose class is taken into account only for verification of the class assignments. In this work, we use the J48 Java based implementation of C4.5 available in the Weka package [68].

Random Forest

Random Forest [69] is an ensemble machine learning method that uses decision trees as basic classifiers. Each tree refers to a class and relies on a random and independent vector, which is generated with the same distribution of the others. Random Forest generates distinct decision trees, because it varies the training sets selection and the selected features for each model. The classification results of a Random Forest execution are computed by counting the votes for the most popular class predicted by the different trees and by assigning that class to the considered instance. Also for Random Forest we use its Weka [68] implementation.

RIPPER

Repeated Incremental Pruning to Produce Error Reduction (RIPPER) [52] is an algorithm based on logic formulas, i.e., a combination of significant features with logic operators (and, or, not, <,≤, ≥, >, =) in the form of “if-then rules”. A rule-based classifier generates a set of rules for assigning a given class to each sample. In this case the features of the sample have to satisfy the conditions of the classification rule for belonging to a given class. Also for this algorithm we use the Weka [68] implementation.

CAMUR

Classifier with Alternative and MUltiple Rule-based models (CAMUR) [51] is a multiple rule-based classifier, which extracts alternative and equivalent classification models. CAMUR combines the RIPPER algorithm with a repeated classification procedure, deleting iteratively the features that appear in the classification models from the dataset in order to extract many classification models. The iterative procedure is stopped when the classification performance is below a given threshold or a given number of iterations has been reached. In this work, we choose a particular execution mode of CAMUR (loose mode), because it allows to extract a larger number of classification models, i.e., more genes that are related to the investigated tumors. Indeed, the loose mode considers all the combinations of the features separately and without definitively removing them from the dataset. For additional details about the algorithm the reader may refer to [51].

Results

In this section, we describe the performed experiments to test our method and the results of the classification algorithms applied to the RNA sequencing and DNA methylation data of three cancer types. We extracted all the samples of these experiments from TCGA, considering the Breast Invasive Carcinoma (BRCA), the Kidney Renal Papillary Cell Carcinoma (KIRP), and the Thyroid Carcinoma (THCA). For data extraction we used the TCGA2BED tool [70] and its TCGA data release in BED format. Every BED file is related to an experiment on a given sample identified by its TCGA barcode [46], which contains several information about the sample including the type. The sample type permits to distinguish between normal and tumoral samples, which are the two classes used for classifying the experiments. For each type of tumor, we created three data matrices, the first containing only the gene expression values (RNA sequencing), the second containing only the gene methylation quantities, and the third combining both experiments according to the procedure described in “Methods” section. Tables 4 and 5 show an example of the RNA sequencing and of the DNA methylation data matrices of BRCA. The numeric values shown in Table 2 are obtained as sums of the beta values associated with the same gene. For example for the TCGA-A7-A4SD-01A-11D-A268-05 sample, the 1.6 value, associated with the GDA gene, is the result of the sum of all the beta values of the methylation sites associated with this gene. Table 6 shows an example of the BRCA combined matrix, where each column represents a gene of the DNA methylation and then a gene of the RNA sequencing; the first row represents a sample only with data about the RNA sequencing experiment, and the second row has only data for the DNA methylation experiment, whereas the last row is an example of a sample with both experiments. Details about the combined matrices of BRCA, THCA, and KIRP tumors are summarized in Table 7, while details about the datasets are depicted in Table 8: with the column ‘Experiment’ we specify the sequencing experiment (RNA sequencing or DNA methylation), followed by the ’Cancer’ column where we indicate with a code the considered types of cancer for the two experiments. The last four columns represent the number of tumor samples, the number of normal samples, the total number of genes and the size of the matrices in MB, respectively. We performed binary classifications (two classes, normal and tumoral), and we considered three cancers with both normal and tumoral samples.

Table 4 Example of RNA sequencing matrix on breast cancer data
Table 5 Example of DNA methylation matrix on breast cancer data
Table 6 Example of combined matrix on the breast cancer data
Table 7 Details of the number of samples in the combined matrices
Table 8 Overview of the datasets

Performed tests

The data matrices of the different experiments and tumors have been analyzed with the above-mentioned classification algorithms (C4.5, Random Forest and RIPPER) through the use of the Weka software package [68]. For the application of these algorithms, we adopted a parameter tuning process to prevent overfitting and to optimize the classification results in term of accuracy. We used the Cross-Validated Parameter selection (CVParameterSelection) [71], that can optimize an arbitrary number of parameters according to input data and number of cross validation folds. We have chosen this meta-classifier for performing parameter selection by cross-validation for all our classifiers. For example, if we consider the RNA-sequencing matrix for KIRP tumor, and the different classifiers (RIPPER, C4.5 and Random Forest), we obtain the following results:

  • J48 (C4.5), -C (confidence threshold for pruning.) 0.1, -M (minimum number of instances per leaf) 1, -U (use unpruned tree) false;

  • JRip (RIPPER), -F (the number of folds for Reduced Error Pruning) 5, -N (the minimal weights of instances within a split) 1, -O (the number of runs of optimizations) 2 -S (the seed of randomization) 1;

  • RandomForest, -I (number of iterations) 30, -K (number of attributes to randomly investigate) 0, -S (seed for random number generator) 1, -num-slots (number of execution slots) 1.

In addition, we performed the classifications with multiple rule-based models obtained by CAMUR. Therefore four different classification algorithms were applied on three data matrices (RNA sequencing, DNA methylation, and their combination) of each considered cancer, resulting in 36 different knowledge discovery analyses. For evaluating the classifiers we take into consideration the F-measure, which is defined as F-\(measure = \frac {2P \cdot R}{P+R}\), where R stands for Recall and P is for Precision. Considering True Positives (TP) objects of a given class recognized in this class; False Positives (FP) objects recognized in a class but not belonging in this class; True Negatives (TN) objects not belonging and not recognized in a given class; False Negatives (FN) objects in a given class but not recognized in that, we can then define \(Recall = \frac {{TP}}{TP+FN}\) and \(Precision = \frac {TP}{TP+FP}\). We performed the tuning of parameters also for CAMUR adopting the Cross-Validated Parameter selection described above [71] for its internal RIPPER algorithm, and we finally set the execution mode to loose, the maximum number of iterations to 100, the minimum F-measure value to 0.8, and the maximum time to 30 days.

In Table 9 we show the average of the resulting F-measures for the performed classifications of each algorithm in 10-fold cross validation scheme. It is worth noting that all values are greater than 95%. Proper parameter tuning was performed with a large set of tests in order to prevent potential overfitting of the classification models.

Table 9 Average performance (F-measure) of the classification algorithms

The results obtained on the combined datasets are slightly lower due to the increase in features and missing values that make the job of the classification algorithms harder. In order to clarify this point we also applied the classification algorithms on the combined matrices, deleting the samples for which only one NGS experiments is available. In this way we reduced the missing values and the resulting classification performance (F-measure) improved with all the classifiers (i.e., on BRCA +0.2% with C4.5, +0.1% with RF, +1% with RIPPER; on KIRP +0.7% with C4.5, +0.8% with RF, +2,1% with RIPPER; on THCA +0.1% with C4.5, +0.5% with RF, +0.2% with RIPPER).

The performance of the algorithms are important in order to validate the classification, but the main purpose of the work is to extract more and different genes from diverse experiments. The improvement given by the classification of the combined data is that the resulting classification models do not only consider the genes and their associated values for a single experiment, but both from gene expression and DNA methylation data in a single model, providing multiple related genes. In Table 10 we show the number of genes obtained with the execution of RIPPER, C4.5 and Random Forest classification algorithms on all tumors for DNA methylation, RNA sequencing and their combination. It is worth noting that one extracted gene is in common among the three algorithms. The reason is that the algorithms operate differently and use diverse extraction functions of the models, so the extracted features are disjoint. It is important to distinguish between Random Forest that extracts multiple classification models, while RIPPER and C4.5 extract a single classification model. We obtain almost 5000 genes with Random Forest, 38 genes with RIPPER, and 26 with C4.5; we also report that 17 genes are in common between RIPPER and Random Forest, 9 between C4.5 and Random Forest, and 4 between RIPPER and C4.5. Further details and the complete list of extracted genes are available as Additional files 1 and 2 or at http://bioinf.iasi.cnr.it/genint.

Table 10 Number of genes obtained with the different classification algorithm

We also investigated if the algorithms misclassify the same samples by comparing the predictions of each one. We found out that only some instances are misclassified by all the three algorithms. Further details are described in Additional file 1.

Finally, in order to prove the validity of the extracted models we performed random permutations of class membership for each classification problem and each combination. We tested if our procedure is able to extract meaningful classification models regardless of the class partition imposed on the training set. This would be verified only in the presence of a marked overfitting behavior. For validating our results and the extracted classification models, we applied the procedure to data with random permutations of class labels. This validation test was performed on 100 different random permutations for each classification problem. In particular, we obtain low values of F-measure and we report the resulting averages in Table 11. We obtain a low overall average classification accuracy on permutated data, whose values are halved when compared to the ones obtained on original data. This confirms the reliability of our classification models and suggesting the absence of overfitting when considering the correct classes.

Table 11 Average performance (F-measure) of the classification algorithms on random permutated class labels

We ran more than 2000 classification procedures with CAMUR, obtaining rules, literal and conjuncion lists, feature pairs and literals statistics for each tumor and each considered dataset. Detailed results are described in Additional file 1 and available at Additional file 2 or at http://bioinf.iasi.cnr.it/genint. In Table 12 we summarize the results obtained with CAMUR, in particular the table shows the total number of extracted rules and all the features (i.e., genes) that appear. In Table 13 we report the execution times, the number of iterations and the execution mode of CAMUR. The execution of the classifications procedures were run on a 4-Core 3 giga hertz Intel-7 processor with 24 gigabytes RAM and Linux Debian Kernel Version 2.6.h-amd64. The classifications obtained with the implementations of C4.5, Random Forest, and RIPPER algorithms, are executed with two software tools available at http://bioinf.iasi.cnr.it/genint. In Table 14 we report the execution times of the classification procedure for each tumor in 10-fold cross-validation sampling scheme [72]. Conversely to CAMUR, the execution times are in the order of minutes, because those algorithm extract just a single classification model. We also compared the execution times of Random Forest to those of CAMUR, which extract both multiple solutions. We note that CAMUR has higher running times than Random Forest, which are in the order of hours for CAMUR and in the order of minutes for Random Forest. We can justify this differences by considering the amount of logic formulas extracted from both classifiers, indeed CAMUR extracts many more rule-based models w.r.t. Random Forest tree-based ones.

Table 12 Rules and genes obtained with CAMUR
Table 13 Timing of the CAMUR executions, number of iterations and execution mode
Table 14 Execution time of C4.5, RIPPER and Random Forest algorithms

Discussion

This section is organized as follows. Firstly, we provide evidence of proof about our new defined measure called gene methylation quantity. Then, we discuss the correlation among RNA sequencing and DNA methylation. Finally, we focus on the extracted knowledge, provide an overview of it, and, in order to prove the validity of the obtained classification models, we considered two external breast cancer RNA sequencing datasets of Gene Expression Omnibus (GSE56022 and GSM1308330) for identifying the diseased samples.

Gene methylation quantity

In previous studies, efforts have been made for aggregating DNA methylation at gene level. In [73] a methylation index is defined as the mean percent methylation across all CpG sites in the gene. In [74] another methylation index is defined as the ratio of methylated and unmethylated copy numbers measured by absolute quantitative assessment of methylated alleles. Our measure differs from previous attempts to represent DNA methylation at gene level, because it takes into account both the number and the values of methylated sites for each gene. In order to validate the gene methylation quantity we provide a qualitative and a quantitative explanation: (i) the defined gene methylation quantity index represents how much a gene is methylated, because it is defined as the sum of the methylation values of the sites that are within the genomic coordinates of the gene, therefore if the gene methylation quantity is low/high, than the gene will be low/high methylated; (ii) we have shown that four classification algorithms are able to successfully distinguish tumoral from non tumoral samples by considering the gene methylation quantities as features. In addition, the index provides a gene oriented data representation of the DNA methylation experiment.

Correlation between DNA methylation and RNA sequencing

An interesting problem is to investigate if there is correlation between gene expression and DNA methylation. The authors of [7580]) address the question if there is correlation between the expression values and the methylated sites of a gene in cancer data and prove that a correlation exists only for a few set of genes. Specifically for the Breast Invasive Carcinoma, in [81] the correlation between DNA methylation and gene expression of almost 3000 genes is discussed, and in [82] it is shown how the CpG-SNP (partnership between DNA methylation and Single Nucleotide Polymorphism) pairs are strongly associated with differential expression of genes. Indeed, DNA methylation has been related also to mutations, and it has been proven that Single Nucleotide Polymorphism at specific loci can result in different patterns of DNA methylation [83].

Tree-based classification models of C4.5

We extracted a classification model for each experiment and each cancer with C4.5, resulting in 9 decision trees composed of 26 genes (16 for DNA methylation and 10 for RNA sequencing). We show some examples on the Kidney Renal Papillary Cell Carcinoma (KIRP) data, in Table 15 we report the RNA sequencing decision tree, and Table 16 shows it for the combined data. The classification models on the other tumors and experiments are described in Additional file 1 and are available at Additional file 2 or http://bioinf.iasi.cnr.it/genint. In the leaves of the trees the total weight of instances reaching that leaf, and the total weight of misclassified instances are specified. In each leaf a fractional weight representing the instances with a missing value is considered. We can then see how missing values are handled by comparing the Tables 15 and 16. In Table 15, the weights of instances are integers, whereas in Table 16 weights are all fractional values, due to the fact that in the combined matrix most of the instances contain missing values. As we can see the model obtained from the combined data provides additional knowledge in the resulting classification rules, compared to that obtained from RNA sequencing data. In particular, the first rule of the model in Table 15 is enriched with additional rule conditions on the genes of the DNA methylation data, as shown in Table 16.

Table 15 The decision tree for full training set, obtained from the RNA sequencing KIRP data matrix, with 319 correctly classified instances and 4 incorrectly classified instances
Table 16 The decision tree for full training set, obtained from the combined KIRP data matrix, with 338 correctly classified instances and 7 incorrectly classified instances

Finally, we validated the tree-based RNA-sequencing classification models on two external datasets extracted from Gene Expression Omnibus (GSE56022 and GSM1308330), obtaining 90% correct classification.

Tree-based classification models of Random Forest

We applied Random Forest to all matrices, extracting 9 classification models, each one composed of 30 trees. The total number of genes obtained is 2301 for RNA sequencing and 2574 for DNA methylation of which 306 are in common between two experiments. Thanks to their combination, we extracted 2471 genes with the execution of this algorithm. As example, we show in Table 17 a random tree obtained with the application of Random Forest on the DNA methylation matrix of Breast Invasive Carcinoma (BRCA) data.

Table 17 model A tree of the classification model for full training set, obtained by the execution of Random Forest on DNA methylation data of Breast Invasive Carcinoma

The Random Forest algorithm is particularly suited for knowledge extraction on combined data (which presents a high number of features), because of its randomized and multiple model extraction.

For the validation of the classification models on the external datasets GSE56022 and GSM1308330, we consider all trees generated for the RNA-sequencing experiment, and the samples are classified with an average accuracy of 80%.

Rule-based classification models of RIPPER

The RIPPER algorithm provides 9 rule-based classification models composed of 38 genes, 22 for DNA methylation and 16 for RNA sequencing. Below we show some examples of the rule-based classification models obtained with the RIPPER algorithm on the Kidney Renal Papillary Cell Carcinoma (KIRP). We show some rules for the DNA methylation dataset in Table 18, and for the combined dataset in Table 19. For example the rule depicted in Table 19 can be interpreted as: classify the considered sample into normal, if the gene methylation quantity of MAP3K11 is lower-equal then 12.3 and the one of PIP5KLI is greater-equal then 2.1 or the RSEM RNA-Seq value of NELL1 is greater-equal then 437.3. Conversely, assign the sample to the tumoral class. The reader may find all the classification models in http://bioinf.iasi.cnr.it/genint.

Table 18 Rule-based model for full training set, obtained from the DNA methylation KIRP data matrix, with 306 correctly classified instances and 11 incorrectly classified instances
Table 19 Rule-based model for full training set, obtained from the combined KIRP data matrix, with 334 correctly classified instances and 11 incorrectly classified instances

The rules in the resulting model obtained from the combined matrix, confirm the added value. In this model we can find the same rule obtained from the single DNA methylation data, enriched with a new rule-based on a feature derived from RNA sequencing data.

We also applied the rule-based RNA-sequencing model extracted on the Gene Expression Omnibus datasets, obtaining a correct classification rate of 90% on average.

Rule-based classification models of CAMUR

By running more than 2000 classification procedures, we extracted 15.252 rules composed of 1758 genes from RNA sequencing and 3655 genes from DNA methylation. From those genes 509 are in common in both experiments. The reader may find the gene lists in Additional file 1 or at http://bioinf.iasi.cnr.it/genint. In this subsection, we show some example of the rules obtained through the execution of CAMUR on the Thyroid Carcinoma (THCA) data. CAMUR extracts many multiple classification models (available at http://bioinf.iasi.cnr.it/genint), as example we report only those with the highest level of accuracy in Table 20.

Table 20 Three classification models shown for full training set, of DNA methylation experiment for Thyroid Carcinoma, with about 100% level of accuracy

The rules for the combined data classification model confirm what is derived from the model for DNA methylation data, and also provide a further classification rule-based on RNA sequencing genes that is not obtained by performing classification on data of single experiment of RNA sequencing. It is worth noting that CAMUR can be successfully adopted for knowledge extraction on combined data (which presents a high number of features), because it is able to extract multiple rule-based models.

For the validation of CAMUR we selected ten BRCA rules extracted by CAMUR and used them on the Gene Expression Omnibus datasets, noting that with these rules 80% of the samples are correctly classified, confirming the validity of the extracted models.

Genes extracted by CAMUR

In this subsection we analyze the large quantity of classification models extracted by CAMUR, focusing on the sets of genes that occur in the rules. We summarize the common genes that appear in the different tumors and experiments with six Venn diagrams, which report the intersections of the genes among the considered tumors and the intersections among each considered experiment.

In Fig. 3 we show the intersections between the tumors sets of genes for the DNA methylation experiment, the RNA sequencing experiment and their combination. The classification models obtained for the DNA methylation experiment of Breast Cancer (BRCA) and of Thyroid Carcinoma (THCA) result in 324 common genes, while the intersections with the sets of genes for the Kidney Renal Carcinoma (KIRP) result in fewer genes (5 for BRCA and KIRP intersection and 6 for THCA and KIRP intersection). Furthermore, 20 genes are in common between all tumors. For RNA sequencing we have 150 genes in common for the BRCA and the THCA tumors. Only 2 genes are extracted for the KIRP tumor that are not in common with the other tumors. In the combined Venn diagram 19 genes are in common between all the tumors, 146 between BRCA and THCA, and less than 10 genes for the intersection of THCA and BRCA with the KIRP set.

Fig. 3
figure 3

Venn diagrams representing the number of genes and their intersection that appear in the DNA methylation experiments, RNA sequencing experiments and in the combined experiments. In the DNA methylation Venn diagram, THCA, BRCA and KIRP sets have 20 common genes, 6 genes are in common between THCA and KIRP, 5 genes between BRCA and KIRP, and 324 between THCA and BRCA. In the RNA sequencing Venn diagram, BRCA and THCA have 150 genes in common, whereas the intersections with the KIRP set of genes are empty, therefore they are not represented.In the combined Venn diagram THCA, BRCA and KIRP sets have 19 common genes, 8 genes are in common between THCA and KIRP, 7 genes between BRCA and KIRP, and 146 between THCA and BRCA

In Fig. 4 we consider a Venn diagram for each tumor, where the set on the left reports the number of genes calculated from the union of genes of RNA sequencing and DNA methylation experiments, and the set on the right stands for the number of genes extracted from the combined matrix. In each tumor different and common genes are extracted from the combined matrices. For example, in BRCA 733 genes are in common, and 312 are the new genes obtained thanks to the combination of RNA sequencing and DNA methylation. We report also 1730 genes that do not appear in the combined dataset of genes. For the THCA tumor 861 gene are in common, 589 belong to the combined dataset of genes, and 1752 genes are extracted from DNA methylation and RNA sequencing matrices. Finally, the KIRP diagram reports 53 new genes for the combined matrix, 35 in common, and 20 present in the union of the two experiments. In this case, the combination produced more genes than those extracted from the single experiments. The combination leads to the extraction of new genes, which are not computed when analyzing single experiments. The goal of this work is also to study the different types of cancer and identify many genes related to the disease. By performing the intersection it is possible to reduce the number of them and to focus on a number of potential oncogenes. In Additional file 2 and at http://bioinf.iasi.cnr.it/genint we provide all the gene lists that are in common and not in common for each tumor and experiment. Thanks to the intersections, we are able to detect 509 genes in common among DNA methylation and RNA sequencing experiments. From these genes we extract a subset of 13 genes, which are in common among the different tumors. In order to check biological relevance of the obtained subset of genes, we compared our result with the Entrez Gene database of NCBI [84], which provides information about oncogenes, tumor suppressor genes, the over-expression or lower-expression of gene regulation of cancer cells growth, and also hyper-methylation and hypo-methylation closely associated with the progression of cancer. This analysis led to the detection of 5 and 279 cancer related genes, from the subsets of 13 and 509 genes discussed above, respectively. Those genes are available as Additional file 2.

Fig. 4
figure 4

The Venn diagrams representing the number of genes and their intersection in each tumor. In KIRP 35 genes are in common between the set of genes extracted from the combined matrix and the union of the genes sets extracted from the DNA methylation and RNA sequencing matrices. We obtain 861 common genes for THCA and 733 for BRCA matrices

Conclusion

In this work, we described the integration of Next Generation Sequencing experiments by distinguishing four cases: (i) integration of data that represent the same NGS experiments, stored in different databases with different schemas; (ii) integration of data that represent different NGS experiments, stored in different databases and with no standardization of schema allowing the access to them; (iii) integration of data that represent different NGS experiments, stored in the same database and with distinct schemas. We also individuated an ideal case for integration (iv), i.e., when a standardization of the schema is provided in order to permit the interoperability between different experiments, with different schemas in different or same databases. We proposed a method for the combination of two distinct NGS experiments with different data schemas. The NGS experiments considered in this study were DNA methylation and RNA sequencing and were extracted from TCGA. We focused on three forms of tumors, i.e. BRCA, KIRP, and THCA. We defined the data matrices, one for each NGS experiment, with samples in the rows and genes in the columns, and a third matrix for representing the combination of DNA methylation and RNA sequencing samples. In particular the objective of the combination was the creation of data matrices indexed on the genes that are related to both NGS experiments. In the RNA sequencing matrices the items are the RNA-Seq by Expectation-Maximization (RSEM) values that quantify the gene expression, whereas in DNA methylation matrices we defined a new measure based on the beta value, that we call gene methylation quantity for denoting the quantity of methylation associates to each gene. After the combination of RNA sequencing and DNA methylation, we proposed the application of supervised analyses. We were able to extract many classification models containing the genes and their quantification values by applying different supervised algorithms (decision trees, rule-based classifiers, and multiple rule-based ones). The classifications were performed on all matrices for each tumor, with the objective to obtain models to separate the normal from the tumoral samples. All the classification models have an accuracy greater than 95%. In particular we obtained 9 decision trees with C4.5, 9 rule-based classification models with RIPPER, and 9 classification models (each one composed of 30 decision trees) with Random Forest. Moreover, thanks to the execution of more than 2000 classification procedures with CAMUR, we extracted 15.252 classification models, from which we derived 5413 genes related to DNA methylation and RNA sequencing. 509 genes are in common among the different experiments and 13 genes among the different tumors. Through the NCBI Entrez gene database we performed functional analysis of those genes. We found 279 out of 509 and 5 out of 13 of them already marked as oncogenes. CAMUR was applied in order to extract possible oncogenes and to find new ones. Many of the extracted genes have been already classified as oncogenes, and this confirms that our method is able to identify relevant genes and justifies further analyses [85]. Indeed, we suggest as future direction a further biological investigation of the classifications models and the extracted sets of genes to confirm their relation with the considered tumors. As other future work, we suggest the application of our method to other forms of tumors. Finally, we plan to define new gene wide measures on different NGS experiments (e.g., mutations, copy number variations, chip-sequencing) in order to consider the combination of them for a comprehensive knowledge extraction.

References

  1. Hayden E. Technology: the $1,000 genome. Nature. 2014; 507(7492):294–5.

    Article  Google Scholar 

  2. Sheridan C. Illumina claims $1,000 genome win. Nat Biotechnol. 2014; 32(2):115.

    Article  Google Scholar 

  3. Weitschek E, Santoni D, Fiscon G, De Cola M, Bertolazzi P, Felici G. Next generation sequencing reads comparison with an alignment-free distance. BMC Res Notes. 2014; 7(1):869.

    Article  Google Scholar 

  4. Jay S, Ji H. Next-generation DNA sequencing. Nat Biotechnol. 2008; 26(10):1135–45.

    Article  Google Scholar 

  5. Mardis E. The impact of next-generation sequencing technology on genetics. Trends Genet. 2008; 24(3):133–41.

    Article  CAS  Google Scholar 

  6. Meldrum C, Doyle M, Tothill R. Next-generation sequencing for cancer diagnostics: a practical perspective. Clin Biochem Rev. 2011; 32(4):177–95.

    PubMed  PubMed Central  Google Scholar 

  7. Koboldt D, Steinberg K, Larson D, Wilson R, Mardis E. The next-generation sequencing revolution and its impact on genomics. Cell. 2013; 155(1):27–38.

    Article  CAS  Google Scholar 

  8. Aravanis A, Lee M, Klausner R. Next-generation sequencing of circulating tumor dna for early cancer detection. Cell. 2017; 168(4):571–4.

    Article  CAS  Google Scholar 

  9. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010; 20(9):1297–303.

    Article  CAS  Google Scholar 

  10. Mortazavi A, Williams B, McCue K, Schaeffer L, Wold B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods. 2008; 5(7):621–8.

    Article  CAS  Google Scholar 

  11. Li B, Dewey C. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011; 12(1):323.

    Article  CAS  Google Scholar 

  12. Yan Z, Cullen B. Sequence requirements for micro RNA processing and function in human cells. RNA. 2003; 9(1):112–23.

    Article  Google Scholar 

  13. Park P. Chip–seq: advantages and challenges of a maturing technology. Nat Rev Genet. 2009; 10(10):669–80.

    Article  CAS  Google Scholar 

  14. Conrad D, Pinto D, Redon R, Feuk L, Gokcumen O, Zhang Y, et al. Origins and functional impact of copy number variation in the human genome. Nature. 2010; 464(7289):704–12.

    Article  CAS  Google Scholar 

  15. Bird A. CpG-rich islands and the function of DNA methylation. Nature. 1985; 321(6067):209–13.

    Article  Google Scholar 

  16. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002; 16(1):6–21.

    Article  CAS  Google Scholar 

  17. Anna P, Manel E. Epigenetic modifications and human disease. Nat Biotechnol. 2010; 28(10):1057–68.

    Article  Google Scholar 

  18. Jones P. Dna methylation and cancer. Cancer Res. 1986; 46(2):461–6.

    CAS  PubMed  Google Scholar 

  19. Ehrlich M. Dna methylation in cancer: too much, but also too little. Oncogene. 2002; 21(35):5400.

    Article  CAS  Google Scholar 

  20. Baylin S. Dna methylation and gene silencing in cancer. Nat Rev Clin Oncol. 2005; 2(S1):S4.

    Article  CAS  Google Scholar 

  21. Li Z, Lei H, Luo M, Wang Y, Dong L, Ma Y, et al. Dna methylation downregulated mir-10b acts as a tumor suppressor in gastric cancer. Gastric Cancer. 2015; 18(1):43–54.

    Article  Google Scholar 

  22. Pickrell J, Marioni J, Pai A, Degner J, Engelhardt B, Nkadori E, et al. Understanding mechanisms underlying human gene expression variation with rna sequencing. Nature. 2010; 464(7289):768–72.

    Article  CAS  Google Scholar 

  23. Eswaran J, Horvath A, Godbole S, Reddy S, Mudvari P, Ohshiro K, et al. Rna sequencing of cancer reveals novel splicing alterations. Sci Rep. 2013; 3:1689.

    Article  Google Scholar 

  24. Min Y, Ting D, Stott S, Wittner B, Ozsolak F, Paul S, et al. Rna sequencing of pancreatic circulating tumour cells implicates wnt signalling in metastasis. Nature. 2012; 487(7408):510–3.

    Article  Google Scholar 

  25. Deng S, Cao S, Huang D, Wang Y. Identifying stages of kidney renal cell carcinoma by combining gene expression and dna methylation data. IEEE/ACM Trans Comput Biol Bioinformatics. 2016; 14(5):1147–1153.

    Article  Google Scholar 

  26. Liggett T, Melnikov A, Tilwalli S, Yi Q, Chen H, Replogle C, et al. Methylation patterns of cell-free plasma dna in relapsing–remitting multiple sclerosis. J Neurol Sci. 2010; 290(1):16–21.

    Article  CAS  Google Scholar 

  27. Toperoff G, Aran D, Kark J, Rosenberg M, Dubnikov T, Nissan B, et al. Genome-wide survey reveals predisposing diabetes type 2-related dna methylation variations in human peripheral blood. Hum Mol Genet. 2012; 21(2):371–83.

    Article  CAS  Google Scholar 

  28. Mill J, Tang T, Kaminsky Z, Khare T, Yazdanpanah S, Bouchard L, et al. Epigenomic profiling reveals dna-methylation changes associated with major psychosis. Am J Hum Genet. 2008; 82(3):696–711.

    Article  CAS  Google Scholar 

  29. Yang X, Gao L, Zhang S. Comparative pan-cancer dna methylation analysis reveals cancer common and specific patterns. Brief Bioinform. 2016; 18(5):761–773.

    Google Scholar 

  30. Zhu Y, Li S, Wang Q, Chen L, Wu K, Huang Y, et al. Quantitative and correlation analysis of the dna methylation and expression of dapk in breast cancer. PeerJ. 2017; e3084:5.

    Google Scholar 

  31. Luk S, Tong M, Ng K, Yip K, Guan X, Ma S. Identification of zfp42/rex1 as a regulator of cancer stemness in cd133+ liver cancer stem cells by genome-wide dna methylation analysis. Nat Genet. 2017; 77(13):4352.

    Google Scholar 

  32. Pan D, Zhang X, Huang C, Jafari N, Kibbe W, Hou L, et al. Comparison of beta-value and m-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics. 2010; 11(1):587.

    Article  Google Scholar 

  33. Handel A, Ebers G, Ramagopalan S. Epigenetics: molecular mechanisms and implications for disease. Trends Mol Med. 2010; 16(1):7–16.

    Article  CAS  Google Scholar 

  34. Trapnell C, Williams B, Pertea G, Mortazavi A, Kwan G, van Baren M, et al. Transcript assembly and quantification by rna-seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010; 28(5):511–5.

    Article  CAS  Google Scholar 

  35. Wang K, Singh D, Zeng Z, Coleman S, Huang Y, Savich G, et al. Mapsplice: accurate mapping of rna-seq reads for splice junction discovery. Nucleic Acids Res. 2010; 38(18):e178.

    Article  Google Scholar 

  36. Wagner G, Kin K, Lynch V. Measurement of mrna abundance using rna-seq data: Rpkm measure is inconsistent among samples. Theory Biosci. 2012; 131(4):281–5.

    Article  CAS  Google Scholar 

  37. Fatih O, Milos P. Rna sequencing: advances, challenges and opportunities. Nat Rev Genet. 2011; 12(2):87–98.

    Article  Google Scholar 

  38. Gomez-Cabrero D, Abugessaisa I, Maier D, Teschendorff A, Merkenschlager M, Gisel A, et al. Data integration in the era of omics: current and future challenges. BMC Syst Biol. 2014; 8(2):I1.

    Article  Google Scholar 

  39. Masseroli M, Kaitoua A, Pinoli P, Ceri S. Modeling and interoperability of heterogeneous genomic big data for integrative processing and querying. Methods. 2016; 111:3–11.

    Article  CAS  Google Scholar 

  40. Endrullat C, Glökler J, Franke P, Frohme M. Standardization and quality management in next-generation sequencing. Appl Transl Genomics. 2016; 10:2–9.

    Article  Google Scholar 

  41. Tatusova T, Karsch-Mizrachi I, Ostell J. Complete genomes in www entrez: data representation and analysis. Bioinformatics. 1999; 15(7):536–43.

    Article  CAS  Google Scholar 

  42. Squizzato S, Park Y, Buso N, Gur T, Cowley A, Li W, et al. The ebi search engine: providing search and retrieval functionality for biological data from embl-ebi. Nucleic Acids Res. 2015; 43(W1):W585–W588.

    Article  CAS  Google Scholar 

  43. Etzold T, Ulyanov A, Argos P. Srs: Information retrieval system for molecular biology data banks. Methods Enzymol. 1996; 266:114–28.

    Article  CAS  Google Scholar 

  44. Freier A, Hofestädt R, Lange M, Scholz U, Stephanik A. Biodataserver: a sql-based service for the online integration of life science data. In silico Biol. 2002; 2(2):37–57.

    CAS  PubMed  Google Scholar 

  45. Zhao Q, Shi X, Xie Y, Huang J, Shia B, Ma S. Combining multidimensional genomic measurements for predicting cancer prognosis: observations from tcga. Brief Bioinformatics. 2015; 16(2):291.

    Article  CAS  Google Scholar 

  46. Weinstein J, Collisson E, Mills G, Shaw K, Ozenberger B, Ellrott K, et al. The cancer genome atlas pan-cancer analysis project. Nat Genet. 2013; 45(10):1113–20.

    Article  Google Scholar 

  47. Esteva A, Kuprel B, Novoa R, Ko J, Swetter S, Blau H, et al. Dermatologist-level classification of skin cancer with deep neural networks. Nature. 2017; 542(7639):115.

    Article  CAS  Google Scholar 

  48. Chen K, Wang K, Tsai M, Wang K, Adrian A, Cheng W, et al. Gene selection for cancer identification: a decision tree model empowered by particle swarm optimization algorithm. BMC Bioinformatics. 2014; 15(1):49.

    Article  Google Scholar 

  49. Pashaei E, Yilmaz A, Ozen M, Aydin N. A novel method for splice sites prediction using sequence component and hidden markov model. In: Engineering in Medicine and Biology Society (EMBC), 2016 IEEE 38th Annual International Conference of the. New York: IEEE: 2016. p. 3076–9.

    Google Scholar 

  50. Li Y, Tang X, Bai Z, Dai X. Exploring the intrinsic differences among breast tumor subtypes defined using immunohistochemistry markers based on the decision tree. Sci Rep. 2016; 6:35773.

    Article  CAS  Google Scholar 

  51. Cestarelli V, Fiscon G, Felici G, Bertolazzi P, Weitschek E. CAMUR: Knowledge extraction from RNA-seq cancer data through equivalent classification rules. Bioinformatics. 2016; 32(5):697–704.

    Article  CAS  Google Scholar 

  52. Cohen W. Fast effective rule induction. In: Proceedings of the twelfth international conference on machine learning. San Francisco: Morgan Kaufmann: 1995. p. 115–23.

    Google Scholar 

  53. Model F, Adorjan P, Olek A, Piepenbrock C. Feature selection for dna methylation based cancer classification. Bioinformatics. 2001; 17(suppl 1):S157—S164.

    Article  Google Scholar 

  54. Bibikova M, Lin Z, Zhou L, Chudin E, Garcia E, Wu B, et al. High-throughput dna methylation profiling using universal bead arrays. Genome Res. 2006; 16(3):383–93.

    Article  CAS  Google Scholar 

  55. Ma X, Wang Y, Zhang M, Gazdar A. Dna methylation data analysis and its application to cancer research. Epigenomics. 2013; 5(3):10.2217/epi.13.26.

    Article  Google Scholar 

  56. Stirzaker C, Taberlay P, Statham A, Clark S. Mining cancer methylomes: prospects and challenges. Trends Genet. 2014; 30(2):75–84.

    Article  CAS  Google Scholar 

  57. Weitschek E, Cumbo F, Cappelli E, Felici G. Genomic data integration: A case study on next generation sequencing of cancer. In: Database and Expert Systems Applications (DEXA), 2016 27th International Workshop on. Los Alamitos: IEEE: 2016. p. 49–53.

    Google Scholar 

  58. Pavlovic M, Ray P, Pavlovic K, Kotamarti A, Chen M, Zhang M. Direction: A machine learning framework for predicting and characterizing dna methylation and hydroxymethylation in mammalian genomes. Bioinformatics. 2017; btx316(btx316). https://doi.org/10.1093/bioinformatics/btx316.

    Article  CAS  Google Scholar 

  59. Celli F, Cumbo F, Weitschek E. Classification of large dna methylation datasets for identifying cancer drivers. Big Data Res. 2018; 13:21–28. https://doi.org/10.1016/j.bdr.2018.02.005.

    Article  Google Scholar 

  60. Golub T, Slonim D, Tamayo P, Huard C, Gaasenbeek M, Mesirov J, et al. Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science. 1999; 286(5439):531–7.

    Article  CAS  Google Scholar 

  61. Khan J, Wei J, Ringner M, Saal L, Ladanyi M, Westermann F, et al. Classification and diagnostic prediction of cancers using gene expression profiling and artificial neural networks. Nat Med. 2001; 7(6):673.

    Article  CAS  Google Scholar 

  62. Sotiriou C, Neo S, McShane L, Korn E, Long P, Jazaeri A, et al. Breast cancer classification and prognosis based on gene expression profiles from a population-based study. Proc Natl Acad Sci. 2003; 100(18):10393–8.

    Article  CAS  Google Scholar 

  63. Weitschek E, Felici G, Bertolazzi P. Mala: a microarray clustering and classification software. In: Database and Expert Systems Applications (DEXA), 2012 23rd International Workshop on Biological Knowledge Discovery. Los Alamitos: IEEE: 2012. p. 201–5.

    Google Scholar 

  64. Marisa L, de Reyniès A, Duval A, Selves J, Gaub M, Vescovo L, et al. Gene expression classification of colon cancer into molecular subtypes: characterization, validation, and prognostic value. PLoS Med. 2013; 10(5):e1001453.

    Article  CAS  Google Scholar 

  65. Novianti P, Roes K, Eijkemans M. Evaluation of gene expression classification studies: factors associated with classification performance. PloS ONE. 2014; 9(4):e96063.

    Article  Google Scholar 

  66. Weitschek E, Fiscon G, Felici G, Bertolazzi P. GELA: A software tool for the analysis of gene expression data. In: 26th International Workshop on Database and Expert Systems Applications, DEXA 2015, Valencia, Spain, September 1-4, 2015. Los Alamitos: 2015. p. 31–35. https://doi.org/10.1109/DEXA.2015.26.

  67. Quinlan J. C4.5: programs for machine learning. San Francisco: Morgan Kaufmann Publishers Inc.; 1993.

    Google Scholar 

  68. Witten I, Frank E, Hall M, Pal C, Vol. 94104. Data Mining: Practical machine learning tools and techniques. 340 Pine Street, 6th Floor San Francisco. USA: Morgan Kaufmann; 2016.

    Google Scholar 

  69. Breiman L. Random forests. Mach Learn. 2001; 45(1):5–32.

    Article  Google Scholar 

  70. Cumbo F, Fiscon G, Ceri S, Masseroli M, Weitschek E. Tcga2bed: extracting, extending, integrating, and querying the cancer genome atlas. BMC Bioinformatics. 2017; 18(1):6.

    Article  Google Scholar 

  71. Kohavi R. Wrappers for performance enhancement and oblivious decision graphs. Tech. rep.Pittsburgh: Carneige-Mellon University Pittsburgh PA, Department of Computer Science; 1995.

    Google Scholar 

  72. Tan P, Steinbach M, Kumar V. Introduction to Data Mining.Boston: Addison Wesley; 2005.

    Google Scholar 

  73. Siegel E, Riggs B, Delmas A, Koch A, Hakam A, Brown K. Quantitative dna methylation analysis of candidate genes in cervical cancer. PLoS ONE. 2015; 10(3):e0122495.

    Article  Google Scholar 

  74. Van Hoesel AQ, Sato Y, Elashoff DA, Turner RR, Giuliano AE, Shamonki JM, et al. Assessment of dna methylation status in early stages of breast cancer development. Br J Cancer. 2013; 108(10):2033.

    Article  CAS  Google Scholar 

  75. Maunakea A, Nagarajan R, Bilenky M, Ballinger T, D’Souza C, Fouse S, et al. Conserved role of intragenic dna methylation in regulating alternative promoters. Nature. 2010; 466(7303):253–7.

    Article  CAS  Google Scholar 

  76. Akalin A, Garrett-Bakelman F, Kormaksson M, Busuttil J, Zhang L, Khrebtukova I, et al. Base-pair resolution dna methylation sequencing reveals profoundly divergent epigenetic landscapes in acute myeloid leukemia. PLoS Genet. 2012; 8(6):e1002781.

    Article  CAS  Google Scholar 

  77. Kulis M, Heath S, Bibikova M, Queirós A, Navarro A, Clot G, et al. Epigenomic analysis detects widespread gene-body dna hypomethylation in chronic lymphocytic leukemia. Nat Genet. 2012; 44(11):1236–42.

    Article  CAS  Google Scholar 

  78. Chen C, Zhang C, Cheng L, Reilly J, Bishop J, Sweeney J, et al. Correlation between dna methylation and gene expression in the brains of patients with bipolar disorder and schizophrenia. Bipolar Disord. 2014; 16(8):790–9.

    Article  CAS  Google Scholar 

  79. Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, Heravi-Moussavi A, et al. Integrative analysis of 111 reference human epigenomes. Nature. 2015; 518(7539):317.

    Article  CAS  Google Scholar 

  80. Li C, Lee J, Ding J, Sun S. Integrative analysis of gene expression and methylation data for breast cancer cell lines. BioData Min. 2018; 11(1):13.

    Article  Google Scholar 

  81. Fleischer T, Frigessi A, Johnson K, Edvardsen H, Touleimat N, Klajic J, et al. Genome-wide dna methylation profiles in progression to in situ and invasive carcinoma of the breast with impact on gene transcription and prognosis. Genome Biol. 2014; 15(8):435.

    PubMed  PubMed Central  Google Scholar 

  82. Shilpi A, Bi Y, Jung S, Patra S, Davuluri R. Identification of genetic and epigenetic variants associated with breast cancer prognosis by integrative bioinformatics analysis. Cancer Informat. 2017; 16:CIN–S39783.

    Article  Google Scholar 

  83. Smith A, Kilaru V, Kocak M, Almli L, Mercer K, Ressler K, et al. Methylation quantitative trait loci (meqtls) are consistently detected across ancestry, developmental stage, and tissue type. BMC Genomics. 2014; 15(1):145.

    Article  Google Scholar 

  84. Maglott D, Ostell J, Pruitt K, Tatusova T. Entrez gene: gene-centered information at ncbi. Nucleic Acids Res. 2005; 33(suppl_1):D54—D58.

    Google Scholar 

  85. Weitschek E, Cumbo F, Cappelli E, Felici G, Bertolazzi P. Classifying Big DNA Methylation Data: A Gene-Oriented Approach. International Conference on Database and Expert Systems Applications. Communications in Computer and Information Science 903 138-149. https://doi.org/10.1007/978-3-319-99133-7.

    Google Scholar 

Download references

Acknowledgements

We wish to thank Riccardo Torlone for supporting this work and the Cineca consortium for assigning us supercomputer resources [grant number HP10CTJZAM] and for providing the PICO infrastructure to carry part of the experiments with the Hadoop YARN cluster.

Funding

This work has been supported by the “nanomax” flagship project (B81J12006270001), by the ERC Advanced Grant GeCo (Datadriven Genomic Computing) [grant number 693174], and by the MoDiag Regione Lazio Project [grant number A0112-2016-13363]. We wish to thank the Cineca consortium for assigning us.

Availability of data and materials

Software and data are available online at http://bioinf.iasi.cnr.it/genint. The original raw data are available at the GDC data portal https://portal.gdc.cancer.gov/.

Author information

Authors and Affiliations

Authors

Contributions

E.W. and G.F. designed research and validated the method. E.C. performed software implementation, data preparation, experimentation, and analysis. E.C. and E.W. wrote the paper. All authors read and approved the final version of this manuscript.

Corresponding author

Correspondence to Emanuel Weitschek.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional files

Additional File 1

Description of the supplementary material. (PDF 99 kb)

Additional File 2

Detailed results. (RAR 6710 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cappelli, E., Felici, G. & Weitschek, E. Combining DNA methylation and RNA sequencing data of cancer for supervised knowledge extraction. BioData Mining 11, 22 (2018). https://doi.org/10.1186/s13040-018-0184-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13040-018-0184-6

Keywords